Las Interacciones Entre La Microbiota y Bacterias Patógenas en El Intestino

You might also like

You are on page 1of 22

HHS Public Access

Author manuscript
Nature. Author manuscript; available in PMC 2016 November 18.
Author Manuscript

Published in final edited form as:


Nature. ; 535(7610): 85–93. doi:10.1038/nature18849.

Interactions between the microbiota and pathogenic bacteria in


the gut
Andreas J. Bäumler1 and Vanessa Sperandio2,3
1Department of Medical Microbiology and Immunology, University of California, Davis, School of
Medicine, Davis, California 95616, USA
2Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas
Author Manuscript

75390-9048, USA
3Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas
75390-9038, USA

Abstract
The microbiome has an important role in human health. Changes in the microbiota can confer
resistance to or promote infection by pathogenic bacteria. Antibiotics have a profound impact on
the microbiota that alters the nutritional landscape of the gut and can lead to the expansion of
pathogenic populations. Pathogenic bacteria exploit microbiota-derived sources of carbon and
nitrogen as nutrients and regulatory signals to promote their own growth and virulence. By
eliciting inflammation, these bacteria alter the intestinal environment and use unique systems for
Author Manuscript

respiration and metal acquisition to drive their expansion. Unravelling the interactions between the
microbiota, the host and pathogenic bacteria will produce strategies for manipulating the
microbiota against infectious diseases.

Appreciation of the important role of the microbiota in human health and nutrition has
grown steadily in the past decade. Initial studies focused on cataloguing the microbial
species that comprise the microbiota and correlating the composition of the microbiota with
the health or disease state of the host. The present period of renaissance has resulted in
technologies and interdisciplinary research that are conducive to mechanistic studies and, in
particular, those that focus on associations between the microbiota, the host and pathogenic
bacteria. Exciting research is now starting to unravel how the composition of the microbiota
can offer either resistance or assistance to invading pathogenic species. The majority of these
Author Manuscript

studies were conducted in the gastrointestinal tract, in which associations between the host
and microbes are of paramount importance. The gut microbiota of each individual is unique
at the genus and species levels; however, it is more generally conserved at the phylum level,
which is populated most prominently by Bacteroidetes and Firmicutes, followed by
Proteobacteria and Actinobacteria. Host genetics, diet and environmental insults such as

Reprints and permissions information is available at www.nature.com/reprints.


Correspondence should be addressed to V.S. (vanessa.sperandio@utsouthwestern.edu).
The authors declare no competing financial interests.
Readers are welcome to comment on the online version of this paper at go.nature.com/28ix4vg.
Bäumler and Sperandio Page 2

treatment with antibiotics alter the microbiota1–4, which can lead to varying susceptibility to
infectious diseases between individuals5.
Author Manuscript

The microbiota can promote resistance to colonization by pathogenic species6–9. For


instance, mice that are treated with antibiotics or that are bred in sterile environments
(known as germ-free mice) are more susceptible to enteric pathogenic bacteria such as
Shigella flexneri, Citrobacter rodentium, Listeria monocytogenes and Salmonella enterica
serovar Typhimurium10–13. And some microbiotas can lead to the expansion or enhanced
virulence of pathogenic populations7. A notable example concerns how differences in the
composition of microbiotas determine the susceptibility of the mice to infection with C.
rodentium: the transplantation of microbiotas from strains of mice that are susceptible to
infection induced similar susceptibility in animals that were previously insusceptible, and
the transplantation of microbiotas from resistant animals led to resistance to infection in
previously susceptible animals14,15. Epidemiological surveys reinforce this idea. For
Author Manuscript

example, differential susceptibility to infection with Campylobacter jejuni was shown to


depend on the species composition of the microbiotas in a study of Swedish adults16.
Individuals with a higher diversity within their microbiotas, and with an abundance of
bacteria from the genera Dorea and Coprococcus, were significantly recalcitrant to C. jejuni
infection compared with people who had low-diversity microbiotas and non-abundance of
Dorea and Coprococcus.

The host’s diet profoundly affects the composition of the microbiota, with repercussions for
the physiology, immunity and susceptibility to infectious diseases of the host17. Dietary
choices have been shown to affect colonization by enterohaemorrhagic Escherichia coli
(EHEC) serotype O157:H7 and the severity and length of its resulting disease18, and
supplementation of the diet with phytonutrients promotes the expansion of beneficial
Author Manuscript

Clostridia species that protect mice from colonization by C. rodentium19.

The use of innovative technologies, in combination with more conventional approaches, is


driving our understanding of the interactions between the microbiota, the host and
pathogenic bacteria. The genetic tractability of several species of bacteria, as well as of their
mammalian hosts (such as mice), allows for the mechanistic investigation of these
relationships. The investigation of changes in the composition of microbiotas has been
driven by next-generation sequencing, which also facilitated the analysis of transcriptomes.
The growing power and finesse of metabolomics studies are quickly expanding our
knowledge of the impact of both the microbiota and of pathogenic bacteria on the metabolic
landscape of the gut. Here, we review advances in our understanding of the complex
relationships that determine the severity and outcome of gastrointestinal infections. The
Author Manuscript

majority of the mechanistic studies that investigate these interactions have been conducted in
S. Typhimurium, EHEC and Clostridium difficile: therefore, these pathogenic organisms are
covered more extensively than others in this Review.

Antibiotics
Antibiotics revolutionized medicine and were justifiably dubbed ‘magic bullets’ against
bacterial infections. However, conventional antibiotics are generally bacteriostatic or

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 3

bactericidal, which means that they indiscriminately kill or prevent the growth of both
Author Manuscript

pathogenic and beneficial microbes. Antibiotics can alter the taxonomic, genomic and
functional features of the microbiota, and their effects can be rapid and sometimes
everlasting20. They can decrease the diversity of the microbiota, which compromises
resistance to colonization by incoming pathogenic bacteria20 — most notably leading to an
expansion of C. difficile that can cause diarrhoea that leads to potentially fatal colitis21.

C. difficile is a spore-forming bacterium that, on germination, colonizes the large intestine


and causes colitis through the action of two toxins: TcdA and TcdB. The majority of C.
difficile infections are nosocomial, but there has also been an increase in community-
acquired infections, mainly due to the ubiquitous presence of C. difficile spores. C. difficile
can colonize the mammalian intestine without causing disease, but one of the most important
risk factors for colitis that is mediated by C. difficile is the use of antibiotics21. The
antibiotics-mediated loss of resistance to colonization also allows colonization by S.
Author Manuscript

Typhimurium and the development of disease22. Both C. difficile and S. Typhimurium


catabolize sialic acid as a source of carbon in the lumen to promote their expansion23. They
rely on saccharolytic members of the microbiota, such as Bacteroides thetaiotaomicron, to
make this sugar freely available in the intestinal lumen. Treatment with antibiotics increases
the abundance of host-derived free sialic acid as well as enhancing its release into the lumen
by B. thetaiotaomicron, which promotes the expansion of the two pathogenic bacteria23.
Antibiotic use also triggers production of the organic acid succinate, another microbiota-
derived nutrient that confers a growth advantage to C. difficile. It is often present at a low
concentration in the microbiotas of conventional mice, but its presence increases on
treatment with antibiotics, which promotes a bloom of C. difficile24 (Fig. 1).

Knowledge of how microbiota disruption affects the ability of bona fide or opportunistic
Author Manuscript

pathogenic organisms to infect hosts is still in its infancy. However, two underlying themes
converge: microbiota-induced changes in the metabolite landscape of the gut and
inflammation.

Utilization of nutrients
Simple dietary sugars are absorbed in the small intestine, which means that they are
unavailable as sources of carbon for the microbiota and pathogenic bacteria in the colon. The
most abundant members of the microbiota are those that are able to utilize the undigested
plant polysaccharides and host glycans that are present in the colon25.

The gut epithelium is protected by a layer of mucus that is composed of proteins known as
mucins that are rich in fucose, galactose, sialic acid, N-acetylgalactosamine, N-
Author Manuscript

acetylglucosamine and mannose. These sugars are harvested by saccharolytic members of


the microbiota, such as Bacteroidales in the gut, which makes them available to species
within the microbiota that lack this capability26. However, pathogenic bacteria in the gut can
also exploit the availability of these sugars to promote their own expansion. Several studies
have used B. thetaiotaomicron as a model Bacteroides in which to investigate these
syntrophic links. Sialic acid is a terminal sugar of some mucosal glycans, and B.
thetaiotaomicron has sialidase activity but lacks the catabolic pathway for sialic-acid

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 4

utilization. The bacterium therefore releases sialic acid to gain access to underlying glycans
Author Manuscript

that it can use as a source of carbon. The sialic acid that B. thetaiotaomicron releases from
the mucus can be catabolized by both C. difficile and S. Typhimurium, which provides them
with a growth advantage23. The ability of the microbiota to use sialic acid therefore depends
on the action of B. thetaiotaomicron, and mutants that lack sialidase fail to enhance the
growth of these two pathogenic bacteria23.

B. thetaiotaomicron also releases fucose from the mucus. It harbours multiple enzymes that
can cleave fucose from host glycans, so its presence results in the high availability of fucose
in the lumen of the gut27–30. This free fucose can also be used as a source of carbon by S.
Typhimurium23. Importantly, B. thetaiotaomicron can promote the fucosylation of mucosal
glycans when introduced into monoassociated germ-free mice31,32.

The microbiota resides in the lumen and the outer mucus layer of the intestine. EHEC,
Author Manuscript

however, aims to achieve a unique niche by closely adhering to the enterocytes of the
intestinal epithelium. To achieve its goal, EHEC must successfully compete with the
microbiota for nutrients. B. thetaiotaomicron does not need to compete with EHEC,
however, because it can utilize polysaccharides; EHEC can only utilize monosaccharides
and disaccharides13,33. EHEC’s main competitors are commensal E. coli, which
preferentially utilizes fucose as a source of carbon when growing in the mammalian
intestine13,33. To circumvent this competition, EHEC utilizes other sources of sugar, such as
galactose, the hexuranates, mannose and ribose, which commensal E. coli cannot catabolize
optimally33,34 (Fig. 2).

EHEC uses fucose as a signalling molecule with which to adjust its metabolism and to
regulate the expression of its virulence repertoire in the lumen and the outer mucus layer of
the colon35. It horizontally acquired a pathogenicity island of genes that encode a fucose-
Author Manuscript

sensing signalling-transduction system35. This system is unique to EHEC and to C.


rodentium35 (which is used extensively in mouse models as a surrogate for the human
pathogen EHEC36). It is composed of the membrane-bound histidine sensor kinase FusK,
which specifically autophosphorylates in response to fucose. FusK then transfers its
phosphate to a response regulator called FusR, which is a transcription factor.
Phosphorylation activates FusR, which represses the expression of the fucose utilization
genes in EHEC, and helps EHEC to avoid the need to compete for this nutrient with
commensal E. coli35. To prevent the unnecessary expenditure of energy by EHEC, FusR
represses the genes that encode the EHEC virulence machinery, a syringe-like apparatus
known as a type III secretion system (T3SS), which the bacterium uses to adhere itself to
enterocytes and highjack the function of these host cells35. EHEC therefore uses fucose, a
Author Manuscript

host-derived signal that is made available by the microbiota, to sense the environment of the
intestinal lumen and to modulate its own metabolism and virulence.

To reach the lining of the epithelium, EHEC and C. rodentium produce mucinases37, which
cleave the protein backbone of mucin-type glycoproteins. Expression of these enzymes is
increased by metabolites that are produced by B. thetaiotaomicron38. Because mucus is one
of the main sources of sugar in the colon, where EHEC and C. rodentium colonize,
obliteration of the mucus layer creates a nutrient-poor environment near the epithelium that

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 5

is referred to as gluconeogenic. The colonization of mice by B. thetaiotaomicron therefore


Author Manuscript

profoundly changes the metabolic landscape of the mouse gut because it raises the levels of
organic acids such as succinate24,38,39. Moreover, several metabolites that indicate a
gluconeogenic environment, such as lactate and glycerate, are also elevated38. EHEC and C.
rodentium sense this gluconeogenic and succinate-rich environment through the
transcriptional regulator Cra. On receiving the cue that they have reached the lining of the
gut epithelium, these bacteria activate the expression of their T3SSs38. EHEC therefore
exploits metabolic cues from B. thetaiotaomicron, and probably other members of the
microbiota, to precisely programme its metabolism and virulence (Fig. 2).

Other pathogenic bacteria can also adjust their gene expression in the presence of
microbiota-produced succinate. C. difficile induces a pathway that converts succinate to
butyrate, which confers a growth advantage in vivo24. Populations of C. difficile mutants
that are unable to convert succinate fail to expand in the gut in the presence of B.
Author Manuscript

thetaiotaomicron24.

Several short-chain fatty acids that are produced by the microbiota, are important
determinants of interactions between the microbiota and pathogenic bacteria in the gut. The
abundance and composition of short-chain fatty acids is distinct in each compartment of the
intestine, and the ability to sense these differences might help pathogenic bacteria in niche
recognition. The most abundant short-chain fatty acids in the gut are acetate, propionate and
butyrate. S. Typhimurium preferably colonizes the ileum40, which generally contains acetate
at a concentration of 30 mM. This acetate concentration enhances the expression of the S.
Typhimurium Salmonella pathogenicity island 1 (SPI-1)-encoded T3SS (T3SS-1), which is
involved in the bacterium’s invasion of the host. Conversely, 70 mM propionate and 20 mM
butyrate, concentrations typical of the colon, suppress the expression of the T3SS-1 (ref. 41).
Author Manuscript

Propionate and butyrate seem to affect the T3SS-1 regulatory cascade at various levels.
However, the detailed mechanism of this regulation is yet to be unravelled. In EHEC,
exposure to the levels of butyrate found in the colon increases the expression of the EHEC
T3SS through post-transcriptional activation of the transcriptional regulator Lrp42. Exposure
to the concentrations of acetate and propionate that are found in the small intestine does not
significantly affect the virulence of EHEC.

Diet has a profound effect on the composition of the microbiota and the concentration of
short-chain fatty acids in the gut17. A diet that is high in fibre results in the enhanced
production of butyrate by the gut microbiota. That increases the host’s expression of
globotriaosylceramide, which is a receptor for the Shiga toxin that is produced by EHEC18.
Shiga toxin can lead to the development of haemolytic uraemic syndrome (HUS) and is the
cause of the morbidity and mortality associated with outbreaks of EHEC43. Consequently,
Author Manuscript

animals that are fed a high-fibre diet are more susceptible to Shiga toxin than are those on a
low-fibre diet and develop more severe disease18. Conversely, increased levels of
microbiota-derived acetate protect animals from disease that is caused by the toxin. Certain
species of Bifidobacteria contribute to higher levels of acetate in the gut, which helps to
improve the barrier function of the intestinal epithelium and to prevent Shiga toxin from
reaching the bloodstream44.

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 6

Enteric pathogenic bacteria also use other nutrients to successfully overcome the
Author Manuscript

microbiota’s resistance to their colonization. Ethanolamine is abundant in the mammalian


intestine45. It can be used as a source of carbon and of nitrogen by a number of pathogenic
species46, and food-borne bacteria are particularly adept at using it. However, it cannot be
metabolized by the majority of commensal species47. S. Typhimurium, EHEC and L.
monocytogenes gain a growth advantage in the intestine through their ability to use this
compound45,48,49. Ethanolamine is also used as a signal by EHEC and S. Typhimurium to
activate the expression of virulence genes50,51. And S. Typhimurium uses hydrogen
produced by the microbiota as an energy source to enhance its growth during the initial stage
of infection52.

The exploitation of microbiota-derived molecules as both nutrients and signals is crucial for
the successful infection of the host by pathogenic bacteria. Although such organisms have
clearly developed many strategies through which to circumvent the microbiota’s resistance
Author Manuscript

to colonization, and in many cases even employ its help, the microbiota pushes back, which
creates an intense competition for resources. The ability of EHEC to colonize the intestine
stems from differences in the sources of sugar that are used by EHEC and by commensal E.
coli. For example, the presence of multiple strains of commensal E. coli with overlapping
nutritional requirements interferes with the colonization of the mouse intestine by EHEC53.
This study uses a streptomycin-treated mouse model of EHEC and three distinct commensal
strains of E. coli to assess differential sugar requirements for the successful colonization of
the intestines53. EHEC could colonize mice that were pre-colonized with any one of the
commensal strains, but it could not colonize mice that were pre-colonized with all three
strains53. EHEC has evolved to exploit distinct sources of sugar during colonization of the
gut. It utilizes catabolic pathways for the hexuronates glucuronate and galacturonate and for
sucrose that are not employed by commensal E. coli within the gut33,53. It can also
Author Manuscript

metabolize several sugars simultaneously. The loss of multiple catabolic pathways has an
additive effect on colonization. This phenomenon is not observed in commensal E. coli,
however, which suggests that E. coli uses available sugars in a stepwise fashion54. EHEC
therefore differs from commensal E. coli in metabolic strategy and the use of nutrients for
the colonization of the mammalian intestine.

C. rodentium is outcompeted and then cleared from the mouse gut through a bloom in the
population of commensal E. coli, which competes with C. rodentium for monosaccharides
for nutrition13. By contrast, C. rodentium is not cleared by B. thetaiotaomicron in germ-free
mice that are fed a diet that contains both monosaccharides, which can be used by
Enterobacteriacae such as C. rodentium, and polysaccharides, which can be used by
Bacteroides. However, when the mice are switched to a diet that consists only of
Author Manuscript

monosaccharides, B. thetaiotaomicron and C. rodentium are forced to compete for sugars,


and B. thetaiotaomicron outcompetes C. rodentium13. The ability of pathogenic bacteria to
successfully compete with commensal species for nutrients is therefore important for their
establishment in the gut.

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 7

Interception of signals from the microbiota and the host


Author Manuscript

The microbiota affects the risks and courses of enteric diseases. Vibrio cholerae is a major
cause of explosive diarrhoea in which there is extensive disruption of the intestinal
population of microbes. Metagenomic studies of the faecal microbiota of people with
cholera in Bangladesh show that recovery is characterized by a certain microbiota signature.
Reconstitution of this microbiota in germ-free mice restricts the infectivity of V. cholerae.
Specifically, the presence of Ruminococcus obeum can hamper the colonization of the
intestines by V. cholerae through the production of the furanone signal autoinducer-2, which
causes the repression of several V. cholerae colonization factors55.

Another example of the effect of microbiota-derived signals on host colonization is their use
by EHEC in the colonization of its ruminal reservoir. EHEC exclusively colonizes the recto–
anal junction of adult cattle. Through the sensor protein SdiA, EHEC detects acyl-
Author Manuscript

homoserine lactone signals from the rumen microbiota, which it uses to reprogram itself to
survive the acidic pH of the animal’s stomachs and to successfully colonize the rectoanal
junction56.

As well as being able to directly detect signals that are derived from the microbiota,
pathogenic bacteria can detect host-derived signals that have been modified by the
microbiota to modulate their virulence. V. cholerae has a type VI secretion system (T6SS),
which it uses to kill other bacteria. During its colonization of the intestine, V. cholerae
comes in contact with the mucosal microbiota, which can affect the composition of bile
acids in the intestine. For example, Bifidobacterium bifidum negatively regulates the T6SS
activity of V. cholerae through the metabolic conversion of three bile acids
(glycodeoxycholic acid, taurodeoxycholic acid and cholic acid) into the bile acid
Author Manuscript

deoxycholic acid. Deoxycholic acid, but not its unmodified salts, decreases the expression of
T6SS genes. This leads to a decrease in the killing of E. coli by V. cholerae owing to bile-
acid conversion by other commensals, which decreases the activity of the T6SS57.

Another microbiota-modified host signal that is detected by pathogenic bacteria is the


neurotransmitter noradrenaline. The gut is highly innervated, and neurotransmitters are
important signals in the gastrointestinal tract, where they modulate peristalsis, the flow of
blood and the secretion of ions58. The microbiota affects the availability of neurotransmitters
in the intestinal lumen, as well as their biosynthesis. For example, the microbiota induces
biosynthesis of serotonin59, and microbiota-derived enzymatic activities increase the levels
of active noradrenaline in the gut lumen60. Noradrenaline is synthesized by the adrenergic
neurons of the enteric nervous system61 and it is inactivated by the host through conjugation
with glucuronic acid (to produce a glucuronide). Microbiota-produced enzymes known as
Author Manuscript

glucuronidases then deconjugate glucuronic acid from noradrenaline, which increases the
amount of active noradrenaline in the lumen of the intestine60. Several pathogenic bacteria
of the gut, including EHEC, S. Typhimurium and V. parahaemolyticus, sense noradrenaline
to activate the expression of virulence genes62–65. Two adrenergic sensors have been
identified in bacteria: the membrane-bound histidine kinases QseC and QseE66,67. QseC also
detects the microbiota-produced signal autoinducer-3 (refs 64 and 66), so the sensing of

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 8

signals from both the host and the microbiota converge at the level of a single receptor, a
Author Manuscript

process known as inter-kingdom signalling.

Inflammation
Although diet and the composition of the microbiota heavily influence the availability of
nutrients in the gut, the host also has an important part to play. A crucial driver of changes in
the gut environment is the inflammatory response of the host. Intestinal inflammation in
people is associated with an imbalance in the microbiota, known as dysbiosis, and is
characterized by a reduced diversity of microbes, a reduced abundance of obligate anaerobic
bacteria and an expansion of facultative anaerobic bacteria in the phylum Proteobacteria,
mostly members of the family Enterobacteriaceae68–73. Similar changes in the composition
of the gut microbiota are observed in mice with chemically induced colitis74 and genetically
induced colitis75. These changes in the structure of the microbiota probably reflect an altered
Author Manuscript

nutritional environment that is created by the inflammatory response of the host.

The availability of nutrients in the large intestine is altered during inflammation through
changes in the composition of mucous carbohydrates. Interleukin (IL)-22, a cytokine that is
prominently induced in the intestinal mucosa when mice and rhesus macaques are infected
with S. Typhimurium76,77, stimulates the epithelial expression of galactoside 2-α-L-
fucosyltransferase 2 and enhances the α(1,2)-fucosylation of mucus carbohydrates78,79. The
gut microbiota can liberate fucose from mucus carbohydrates23,80, which leads to the
induction of genes for fucose utilization in E. coli78. Similarly, increased fucosylation of
glycans is observed during S. Typhimurium-induced colitis in mice, which correlates with
elevated synthesis of the proteins involved in fucose utilization81. Mucus fucosylation that is
induced during infection with C. rodentium causes changes in the composition of the gut
Author Manuscript

microbiota that help to protect the host from the expansion and epithelial translocation of the
pathobiont Enterococcus faecalis79.

Another driver of changes in the nutritional environment of the gut is the generation of
reactive oxygen species and reactive nitrogen species during inflammation. Pro-
inflammatory cytokines such as interferon-γ (IFN-γ) activate dual oxidase 2 in the
intestinal epithelium, which produces hydrogen peroxide82. Increased expression of
DUOX2, the gene that encodes dual oxidase 2, in the intestinal mucosa of patients with
Crohn’s disease and ulcerative colitis correlates with an expansion of Proteobacteria in the
gut microbiota83. IFN-γ also induces epithelial expression of the gene Nos2 (ref. 84), which
encodes inducible nitric oxide synthase, the enzyme that catalyses the production of nitric
oxide from L-arginine85. As a result, the concentration of nitric oxide is elevated in gases
from the colons of people with inflammatory bowel disease86–88. Although reactive oxygen
Author Manuscript

and nitrogen species have antimicrobial activity, these radicals quickly form non-toxic
compounds in the lumen of the gut as they diffuse away from the epithelium. For example,
when they are generated during inflammation by host enzymes in the intestinal epithelium,
these species react to form nitrate89. This by-product of inflammation is present at elevated
concentrations in the intestines of mice with chemically induced colitis90 (Fig. 3). Nitrate
reductases, enzymes that are broadly conserved among the Enterobacteriaceae, couple the
reduction of nitrate to energy-conserving electron transport systems for respiration, a process

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 9

termed nitrate respiration. However, the genes that encode them are absent from the
genomes of obligate anaerobic Clostridia or Bacteroidia91. Nitrate respiration drives the
Author Manuscript

Nos2-dependent expansion of commensal E. coli in mice with chemically or genetically


induced colitis, but not in animals without signs of intestinal inflammation91. Respiratory
electron acceptors that are generated as a by-product of the host inflammatory response
therefore create a niche in the lumen of the intestines that supports the uncontrolled
expansion of commensal Enterobacteriaceae rather than of obligate anaerobic bacteria91.
The resulting bloom in the inflamed intestine is one of the most consistent and robust
ecological patterns that has been observed in the gut microbiota92.

The creation of a niche for respiratory nutrients during inflammation is also an important
driver of the strategies that pathogenic bacteria from the family Enterobacteriaceae use to
invade the gut ecosystem. In the absence of inflammation or treatment with antibiotics,
members of the gut microbiota occupy all available nutrient niches, which makes it very
Author Manuscript

challenging for pathogenic Enterobacteriaceae to enter the community. One solution is for
these bacteria to trigger intestinal inflammation, which would coerce the host into creating a
fresh niche of respiratory nutrients that is suitable for its expansion — an approach that is
used by S. Typhimurium93. On ingestion, S. Typhimurium uses T3SS-1 to invade the
intestinal epithelium94 and T3SS-2 to survive in the tissue of the host95. Both of these
processes trigger acute intestinal inflammation in cattle and in mouse models of
gastroenteritis96–98 (Fig. 3). The inflammatory response of the host drives the expansion of
S. Typhimurium in the lumen of the gut99, which is required for the transmission of this
pathogenic species to a new host through the faecal–oral route100.

Although such expansion allows S. Typhimurium to side-step competition with obligate


anaerobic Clostridia and Bacteroidia, this strategy forces the bacterium into battle with
Author Manuscript

commensal Enterobacteriaceae over limited resources. For example, S. Typhimurium


expands in the inflamed gut through nitrate respiration101,102, which results in rivalry with
commensal Enterobacteriaceae that pursue a similar strategy91. S. Typhimurium can gain an
edge in this competition through its ability to utilize a broader range of inflammation-
derived electron acceptors than its rivals. A source of one such electron acceptor is sulfate-
reducing species of Desulfovibrio from the microbiota, which release hydrogen sulfide, a
compound that is converted to thiosulfate by the epithelium of the colon to avoid toxicity103.
Deployment of the virulence factors of pathogenic bacteria leads to the recruitment of
neutrophils to the intestinal mucosa, which is the histopathological hallmark of S.
Typhimurium-induced gastroenteritis96. A fraction of these recruited neutrophils migrate
into the lumen of the intestine — a diagnostic marker of inflammatory diarrhoea104. In the
lumen, neutrophils help to protect the mucosa by engulfing bacteria in the vicinity of the
Author Manuscript

epithelium105, but reactive oxygen species that are generated by the phagocyte-produced
NADPH oxidase 2 (also known as cytochrome b-245 heavy chain) convert thiosulfate into
tetrathionate, a respiratory electron acceptor that supports the expansion of S. Typhimurium
in the lumen of the inflamed gut106 (Fig. 3). Although tetrathionate respiration is a
characteristic of Salmonella serovars and has been used empirically in their isolation in
clinical microbiology laboratories since 1923 (ref. 107), insights into the respiratory nutrient
niche that Salmonella occupies suggest that this property is part of a strategy to edge out
competing commensal Enterobacteriaceae in the inflamed gut106.

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 10

The inflammatory response of the host also ignites competition between commensal and
Author Manuscript

pathogenic Enterobacteriaceae over trace elements such as iron, which is less available
during inflammation. IL-22 induces the release of the antimicrobial protein lipocalin-2 (also
known as neutrophil gelatinase-associated lipocalin) from the epithelium in mice and rhesus
macaques108,109. Lipocalin-2 reduces iron availability by binding to enterobactin, a low-
molecular-weight iron chelator (or siderophore) that is produced by
Enterobacteriaceae110,111. To overcome this, S. Typhimurium and some commensal E. coli
secrete a glycosylated derivative of enterobactin, termed salmochelin, which is not bound by
lipocalin-2 (ref. 108). By producing salmochelin as well as two further siderophores that are
not bound by lipocalin-2, yersiniobactin and aerobactin, the probiotic E. coli strain Nissle
1917 can limit the expansion of S. Typhimurium in the lumen of the inflamed gut112.
Conversely, lipocalin-2 secretion by the epithelium generates an environment that enables S.
Typhimurium to edge out commensal Enterobacteriaceae that depend solely on enterobactin
for the acquisition of iron109 (Fig. 3).
Author Manuscript

Through its limitation of iron availability, intestinal inflammation also sets the stage for
battles between Enterobacteriaceae that use protein-based toxins known as colicins113 that
affect a narrow range of hosts. Iron limitation induces the synthesis of siderophore receptor
proteins for the bacterial outer membrane113, which also commonly serve as receptors for
colicins114–116. Expression of a siderophore receptor protein termed the colicin I receptor
(CirA) confers commensal E. coli with sensitivity to colicin Ib produced by S.
Typhimurium113. The respiratory nutrient niche that is generated by the inflammatory
response of the host is therefore a battleground on which commensal and pathogenic
Enterobacteriaceae struggle for dominance using a diverse arsenal of nutritional and
antimicrobial strategies.
Author Manuscript

Perspective and the future


The study of the microbiome began more than a century ago. equencing of 16S rRNA genes
provided the first insights into the taxonomic composition of microbial communities. Later,
sequencing of the complete metagenome of microbial communities provided a more detailed
insight into the full genetic capacity of such a community. The use of germ-free animals,
either alone or in combination with emerging technologies such as laser-capture
microdissection and transcriptomics, enabled mechanistic studies of the associations
between the microbiota, the host and pathogenic bacteria117. Multi-taxon insertion
sequencing now allows researchers to investigate both the assembly and the shared and
strain-specific dietary requirements of communities of microbes, and it has also facilitated
the informed manipulation of such communities through diet118. The development of
Author Manuscript

quantitative imaging technologies has provided insight into the localization of microbes
within the gastrointestinal tract, and it has also enabled studies on the proximity of and
interactions between microbes119. The increasing refinement and power of metabolomics,
imaging mass spectrometry and three-dimensional mapping of mass-spectrometry data
provide a high-resolution image of the complex chemistry landscape of the interactions
between microbes and the host, which sets the stage for manipulating this chemistry to
prevent or treat infectious diseases24,38,120–127. A marriage of metagenomics and
mathematical modelling promises to enhance the precision of microbiome reconstitution,

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 11

which has proven successful for tackling C. difficile infections in mice128. In these exciting
Author Manuscript

times, the expansion of multidisciplinary research is rapidly generating new technologies


and mechanistic insights into interactions between the microbiota, the host and pathogenic
bacteria (Box 1).

BOX 1

Microbiota interventions as therapeutic strategies to limit pathogen


expansion
An imbalance in the gut microbiota might underlie many human diseases but, in most
cases, the development of treatment options is still in its infancy. This could be in part
because the mechanisms that lead to adverse effects in the host differ for each disease,
which means that intervention strategies must be developed for each. The treatment
options for antibiotic-induced dysbiosis are perhaps the most advanced, mainly because
Author Manuscript

faecal microbiota transplantation can reverse this imbalance in the gut microbiota129.
Nonetheless, the mechanisms through which treatment with antibiotics encourages an
uncontrolled expansion of the obligate anaerobe C. difficile differ markedly from those
that stimulate the growth of the facultative anaerobes Enterobacteriaceae, which has
implications for the development of precision microbiome interventions.

Mice that are treated with streptomycin have a reduced abundance of members of the
class Clostridia130, which are credited with producing the lion’s share of the short-chain
fatty acid butyrate in the large intestine131. The resulting depletion of short-chain fatty
acids drives an expansion of Enterobacteriaceae through mechanisms that are not fully
resolved44,132. Depletion of Clostridia-derived butyrate affects the metabolism of
enterocytes in the colon, which derive most of their energy by butyrate respiration133.
Author Manuscript

The depletion of short-chain fatty acids also leads to a contraction in the pool of
regulatory T cells in the colonic mucosa134–136. These changes in the host physiology
increase the inflammatory tone of the mucosa, as indicated by the elevated expression of
Nos2, the gene that encodes inducible nitric oxide synthase, and contributes to the
expansion of commensal E. coli through nitrate generation137. Although other
mechanisms probably contribute to the post-antibiotic expansion of certain populations of
bacteria in the gut126, the transfer of Clostridia, with their capacity for producing short-
chain fatty acids, represents the most effective treatment for limiting the growth of E. coli
in streptomycin-treated mice138.

By contrast, the post-antibiotic expansion of the C. difficile population is driven by a


depletion of secondary bile salts. The liver produces the primary bile salts cholate and
chenodeoxycholate, which are conjugated to the amino acids taurine (to produce
Author Manuscript

taurocholate and taurochenodeoxycholate) or glycine (to produce glycocholate and


glycochenodeoxycholate) and then secreted into the gut. Bile salt hydrolases, enzymes
that are produced by many members of the gut microbiota, remove the conjugated amino
acid from the primary bile salt. C. scindens is one of a limited number of species of
bacteria that can actively transport cholate and chenodeoxycholate into its cytosol, where
these unconjugated primary bile salts are converted into the secondary bile salts
deoxycholate and lithocholate, which are subsequently secreted into the extracellular

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 12

environment139 (Box Fig.). Although both primary and secondary bile salts induce the
Author Manuscript

germination of C. difficile spores, only secondary bile salts efficiently prevent the growth
of vegetative C. difficile cells140. By significantly reducing the abundance of species that
are capable of producing deoxycholate and lithocholate, treatment with antibiotics causes
a depletion of these secondary bile salts and promotes the expansion of vegetative C.
difficile cells in the large intestine141,142. Faecal microbiota transplantation restores the
production of secondary bile salts and therefore prevents the expansion of C. difficile143.
Direct supplementation of the diet with secondary bile salts warrants caution because
increased concentrations of bile salts have been linked to gastrointestinal cancers144.
However, inoculation with only the secondary-bile-salt-producing C. scindens confers
mice with resistance to C. difficile expansion following treatment with antibiotics128.
This remarkable observation opens the door to novel precision microbiome interventions
that aim to prevent or treat the colitis that is associated with C. difficile infection after
Author Manuscript

antibiotic therapy.
Author Manuscript

Acknowledgments
Work in the V.S. laboratory is supported by US National Institutes of Health (NIH) grants AI053067, AI077613,
AI05135 and AI114511. Work in the A.J.B. laboratory is supported by US Department of Agriculture grant
Author Manuscript

2015-67015-22930 and NIH grants AI044170, AI096528, AI112445, AI114922 and AI117940. The contents of this
Review are solely the responsibility of the authors and do not necessarily represent the official views of the NIH
National Institute of Allergy and Infectious Diseases.

References
1. Eckburg PB, et al. Diversity of the human intestinal microbial flora. Science. 2005; 308:1635–1638.
[PubMed: 15831718]

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 13

2. Frank DN, et al. Molecular-phylogenetic characterization of microbial community imbalances in


human inflammatory bowel diseases. Proc Natl Acad Sci USA. 2007; 104:13780–13785. [PubMed:
Author Manuscript

17699621]
3. The Human Microbiome Project Consortium. Structure function and diversity of the healthy human
microbiome. Nature. 2012; 486:207–214. [PubMed: 22699609]
4. Ley RE, Peterson DA, Gordon JI. Ecological and evolutionary forces shaping microbial diversity in
the human intestine. Cell. 2006; 124:837–848. [PubMed: 16497592]
5. Yurist-Doutsch S, Arrieta MC, Vogt SL, Finlay BB. Gastrointestinal microbiota-mediated control of
enteric pathogens. Annu Rev Genet. 2014; 48:361–382. [PubMed: 25251855]
6. Sassone-Corsi M, Raffatellu M. No vacancy: how beneficial microbes cooperate with immunity to
provide colonization resistance to pathogens. J Immunol. 2015; 194:4081–4087. [PubMed:
25888704]
7. Cameron EA, Sperandio V. Frenemies: signaling and nutritional integration in pathogen–
microbiota–host interactions. Cell Host Microbe. 2015; 18:275–284. [PubMed: 26355214]
8. Pacheco AR, Sperandio V. Enteric pathogens exploit the microbiota-generated nutritional
environment of the gut. Microbiol Spectr. 2015; 3 MBP-0001–2014.
Author Manuscript

9. Bohnhoff M, Drake BL, Miller CP. Effect of streptomycin on susceptibility of intestinal tract to
experimental Salmonella infection. Proc Soc Exp Biol Med. 1954; 86:132–137. [PubMed:
13177610]
10. Ferreira RB, et al. The intestinal microbiota plays a role in Salmonella-induced colitis independent
of pathogen colonization. PLoS ONE. 2011; 6:e20338. [PubMed: 21633507]
11. Sprinz H, et al. The response of the germfree guinea pig to oral bacterial challenge with
Escherichia coli and Shigella flexneri. Am J Pathol. 1961; 39:681–695. [PubMed: 13915950]
12. Zachar Z, Savage DC. Microbial interference and colonization of the murine gastrointestinal tract
by Listeria monocytogenes. Infect Immun. 1979; 23:168–174. [PubMed: 106003]
13. Kamada N, et al. Regulated virulence controls the ability of a pathogen to compete with the gut
microbiota. Science. 2012; 336:1325–1329. This study showed that enteric pathogens use
virulence genes to compete with the microbiota for nutrients in the gut. [PubMed: 22582016]
14. Ghosh S, et al. Colonic microbiota alters host susceptibility to infectious colitis by modulating
inflammation, redox status, and ion transporter gene expression. Am J Physiol Gastrointest Liver
Author Manuscript

Physiol. 2011; 301:G39–G49. [PubMed: 21454446]


15. Willing BP, Vacharaksa A, Croxen M, Thanachayanont T, Finlay BB. Altering host resistance to
infections through microbial transplantation. PLoS ONE. 2011; 6:e26988. [PubMed: 22046427]
16. Kampmann C, Dicksved J, Engstrand L, Rautelin H. Composition of human faecal microbiota in
resistance to Campylobacter infection. Clin Microbiol Infect. 2016; 22:61.e1–61.e8. [PubMed:
26369602]
17. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome
and the immune system. Nature. 2011; 474:327–336. [PubMed: 21677749]
18. Zumbrun SD, et al. Dietary choice affects Shiga toxin-producing Escherichia coli (STEC)
O157:H7 colonization and disease. Proc Natl Acad Sci USA. 2013; 110:E2126–E2133. [PubMed:
23690602]
19. Wlodarska M, Willing BP, Bravo DM, Finlay BB. Phytonutrient diet supplementation promotes
beneficial Clostridia species and intestinal mucus secretion resulting in protection against enteric
infection. Sci Rep. 2015; 5:9253. [PubMed: 25787310]
20. Modi SR, Collins JJ, Relman DA. Antibiotics and the gut microbiota. J Clin Invest. 2014;
Author Manuscript

124:4212–4218. [PubMed: 25271726]


21. Leffler DA, Lamont JT. Clostridium difficile infection. N Engl J Med. 2015; 373:287–288.
22. Pavia AT, et al. Epidemiologic evidence that prior antimicrobial exposure decreases resistance to
infection by antimicrobial-sensitive Salmonella. J Infect Dis. 1990; 161:255–260. [PubMed:
2299207]
23. Ng KM, et al. Microbiota-liberated host sugars facilitate post-antibiotic expansion of enteric
pathogens. Nature. 2013; 502:96–99. This study showed that treatment with antibiotics enhances
the abundance of host sialic acid that can be harvested by the microbiota, which promotes the
expansion of enteric pathogens. [PubMed: 23995682]

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 14

24. Ferreyra JA, et al. Gut microbiota-produced succinate promotes C. difficile infection after
antibiotic treatment or motility disturbance. Cell Host Microbe. 2014; 16:770–777. [PubMed:
Author Manuscript

25498344]
25. Ferreyra JA, Ng KM, Sonnenburg JL. The enteric two-step: nutritional strategies of bacterial
pathogens within the gut. Cell Microbiol. 2014; 16:993–1003. [PubMed: 24720567]
26. Rakoff-Nahoum S, Coyne MJ, Comstock LE. An ecological network of polysaccharide utilization
among human intestinal symbionts. Curr Biol. 2014; 24:40–49. [PubMed: 24332541]
27. Alverdy J, Chi HS, Sheldon GF. The effect of parenteral nutrition on gastrointestinal immunity.
The importance of enteral stimulation. Ann Surg. 1985; 202:681–684. [PubMed: 3935061]
28. Fischbach MA, Sonnenburg JL. Eating for two: how metabolism establishes interspecies
interactions in the gut. Cell Host Microbe. 2011; 10:336–347. [PubMed: 22018234]
29. Chow WL, Lee YK. Free fucose is a danger signal to human intestinal epithelial cells. Br J Nutr.
2008; 99:449–454. [PubMed: 17697405]
30. Bourlioux P, Koletzko B, Guarner F, Braesco V. The intestine and its microflora are partners for the
protection of the host: report on the Danone Symposium “The Intelligent Intestine,” held in Paris,
June 14, 2002. Am J Clin Nutr. 2003; 78:675–683. [PubMed: 14522724]
Author Manuscript

31. Bry L, Falk PG, Midtvedt T, Gordon JI. A model of host–microbial interactions in an open
mammalian ecosystem. Science. 1996; 273:1380–1383. [PubMed: 8703071]
32. Hooper LV, Xu J, Falk PG, Midtvedt T, Gordon JI. A molecular sensor that allows a gut
commensal to control its nutrient foundation in a competitive ecosystem. Proc Natl Acad Sci USA.
1999; 96:9833–9838. [PubMed: 10449780]
33. Fabich AJ, et al. Comparison of carbon nutrition for pathogenic and commensal Escherichia coli
strains in the mouse intestine. Infect Immun. 2008; 76:1143–1152. [PubMed: 18180286]
34. Autieri SM, et al. L-fucose stimulates utilization of D-ribose by Escherichia coli MG1655 ΔfucAO
and E. coli Nissle 1917 ΔfucAO mutants in the mouse intestine and in M9 minimal medium. Infect
Immun. 2007; 75:5465–5475. [PubMed: 17709419]
35. Pacheco AR, et al. Fucose sensing regulates bacterial intestinal colonization. Nature. 2012;
492:113–117. This paper showed that sugars from the mucus that are released by the microbiota
can be perceived as signals by enteric pathogens to regulate the expression of virulence genes.
[PubMed: 23160491]
Author Manuscript

36. Schauer DB, Falkow S. Attaching and effacing locus of a Citrobacter freundii biotype that causes
transmissible murine colonic hyperplasia. Infect Immun. 1993; 61:2486–2492. [PubMed:
8500884]
37. Szabady RL, Lokuta MA, Walters KB, Huttenlocher A, Welch RA. Modulation of neutrophil
function by a secreted mucinase of Escherichia coli O157:H7. PLoS Pathog. 2009; 5:e1000320.
[PubMed: 19247439]
38. Curtis MM, et al. The gut commensal Bacteroides thetaiotaomicron exacerbates enteric infection
through modification of the metabolic landscape. Cell Host Microbe. 2014; 16:759–769. This
study revealed that metabolites that are produced by the microbiota can be exploited as cues to
increase the virulence of enteric pathogens. [PubMed: 25498343]
39. Macy JM, Ljungdahl LG, Gottschalk G. Pathway of succinate and propionate formation in
Bacteroides fragilis. J Bacteriol. 1978; 134:84–91. [PubMed: 148460]
40. Carter PB, Collins FM. The route of enteric infection in normal mice. J Exp Med. 1974; 139:1189–
1203. [PubMed: 4596512]
41. Lawhon SD, Maurer R, Suyemoto M, Altier C. Intestinal short-chain fatty acids alter Salmonella
Author Manuscript

typhimurium invasion gene expression and virulence through BarA/SirA. Mol Microbiol. 2002;
46:1451–1464. [PubMed: 12453229]
42. Takao M, Yen H, Tobe T. LeuO enhances butyrate-induced virulence expression through a positive
regulatory loop in enterohaemorrhagic Escherichia coli. Mol Microbiol. 2014; 93:1302–1313.
[PubMed: 25069663]
43. Karmali MA, Petric M, Lim C, Fleming PC, Steele BT. Escherichia coli cytotoxin, haemolytic-
uraemic syndrome, and haemorrhagic colitis. Lancet. 1983; 2:1299–1300.
44. Fukuda S, et al. Bifidobacteria can protect from enteropathogenic infection through production of
acetate. Nature. 2011; 469:543–547. This study showed that the acetate that is produced by certain

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 15

probiotic bacteria can enhance the barrier function of the gut epithelium to protect the host from
enteric infections. [PubMed: 21270894]
Author Manuscript

45. Bertin Y, et al. Enterohaemorrhagic Escherichia coli gains a competitive advantage by using
ethanolamine as a nitrogen source in the bovine intestinal content. Environ Microbiol. 2011;
13:365–377. [PubMed: 20849446]
46. Garsin DA. Ethanolamine utilization in bacterial pathogens: roles and regulation. Nature Rev
Microbiol. 2010; 8:290–295. [PubMed: 20234377]
47. Korbel JO, et al. Systematic association of genes to phenotypes by genome and literature mining.
PLoS Biol. 2005; 3:e134. [PubMed: 15799710]
48. Thiennimitr P, et al. Intestinal inflammation allows Salmonella to use ethanolamine to compete
with the microbiota. Proc Natl Acad Sci USA. 2011; 108:17480–17485. [PubMed: 21969563]
49. Joseph B, et al. Identification of Listeria monocytogenes genes contributing to intracellular
replication by expression profiling and mutant screening. J Bacteriol. 2006; 188:556–568.
[PubMed: 16385046]
50. Kendall MM, Gruber CC, Parker CT, Sperandio V. Ethanolamine controls expression of genes
encoding components involved in interkingdom signaling and virulence in enterohemorrhagic
Author Manuscript

Escherichia coli O157:H7. mBio. 2012; 3:00050–12.


51. Anderson CJ, Clark DE, Adli M, Kendall MM. Ethanolamine signaling promotes Salmonella niche
recognition and adaptation during infection. PLoS Pathog. 2015; 11:e1005278. [PubMed:
26565973]
52. Maier L, et al. Microbiota-derived hydrogen fuels Salmonella Typhimurium invasion of the gut
ecosystem. Cell Host Microbe. 2013; 14:641–651. [PubMed: 24331462]
53. Maltby R, Leatham-Jensen MP, Gibson T, Cohen PS, Conway T. Nutritional basis for colonization
resistance by human commensal Escherichia coli strains HS and Nissle 1917 against E. coli
O157:H7 in the mouse intestine. PLoS ONE. 2013; 8:e53957. [PubMed: 23349773]
54. Fabich AJ, et al. Comparison of carbon nutrition for pathogenic and commensal Escherichia coli
strains in the mouse intestine. Infect Immun. 2008; 76:1143–1152. [PubMed: 18180286]
55. Hsiao A, et al. Members of the human gut microbiota involved in recovery from Vibrio cholerae
infection. Nature. 2014; 515:423–426. [PubMed: 25231861]
56. Hughes DT, et al. Chemical sensing in mammalian host–bacterial commensal associations. Proc
Author Manuscript

Natl Acad Sci USA. 2010; 107:9831–9836. [PubMed: 20457895]


57. Bachmann V, et al. Bile salts modulate the mucin-activated type VI secretion system of pandemic
Vibrio cholerae. PLoS Negl Trop Dis. 2015; 9:e0004031. [PubMed: 26317760]
58. Hörger S, Schultheiss G, Diener M. Segment-specific effects of epinephrine on ion transport in the
colon of the rat. Am J Physiol. 1998; 275:G1367–G1376. [PubMed: 9843774]
59. Yano JM, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis.
Cell. 2015; 161:264–276. [PubMed: 25860609]
60. Asano Y, et al. Critical role of gut microbiota in the production of biologically active, free
catecholamines in the gut lumen of mice. Am J Physiol Gastrointest Liver Physiol. 2012;
303:G1288–G1295. [PubMed: 23064760]
61. Furness JB. Types of neurons in the enteric nervous system. J Auton Nerv Syst. 2000; 81:87–96.
[PubMed: 10869706]
62. Curtis MM, Sperandio V. A complex relationship: the interaction among symbiotic microbes,
invading pathogens, and their mammalian host. Mucosal Immunol. 2011; 4:133–138. [PubMed:
21248724]
Author Manuscript

63. Moreira CG, Weinshenker D, Sperandio V. QseC mediates Salmonella enterica serovar
Typhimurium virulence in vitro and in vivo. Infect Immun. 2010; 78:914–926. [PubMed:
20028809]
64. Sperandio V, Torres AG, Jarvis B, Nataro JP, Kaper JB. Bacteria–host communication: the
language of hormones. Proc Natl Acad Sci USA. 2003; 100:8951–8956. This paper describes how
signals from the microbiota and host converge to enhance virulence in enteric pathogens.
[PubMed: 12847292]

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 16

65. Nakano M, Takahashi A, Sakai Y, Nakaya Y. Modulation of pathogenicity with norepinephrine


related to the type III secretion system of Vibrio parahaemolyticus. J Infect Dis. 2007; 195:1353–
Author Manuscript

1360. [PubMed: 17397007]


66. Clarke MB, Hughes DT, Zhu C, Boedeker EC, Sperandio V. The QseC sensor kinase: a bacterial
adrenergic receptor. Proc Natl Acad Sci USA. 2006; 103:10420–10425. [PubMed: 16803956]
67. Reading NC, Rasko DA, Torres AG, Sperandio V. The two-component system QseEF and the
membrane protein QseG link adrenergic and stress sensing to bacterial pathogenesis. Proc Natl
Acad Sci USA. 2009; 106:5889–5894. [PubMed: 19289831]
68. Seksik P, et al. Alterations of the dominant faecal bacterial groups in patients with Crohn’s disease
of the colon. Gut. 2003; 52:237–242. [PubMed: 12524406]
69. Gophna U, Sommerfeld K, Gophna S, Doolittle WF, Veldhuyzen van Zanten SJ. Differences
between tissue-associated intestinal microfloras of patients with Crohn’s disease and ulcerative
colitis. J Clin Microbiol. 2006; 44:4136–4141. [PubMed: 16988016]
70. Baumgart M, et al. Culture independent analysis of ileal mucosa reveals a selective increase in
invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn’s
disease involving the ileum. ISME J. 2007; 1:403–418. [PubMed: 18043660]
Author Manuscript

71. Walker AW, et al. High-throughput clone library analysis of the mucosaassociated microbiota
reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in
inflammatory bowel disease. BMC Microbiol. 2011; 11:7. [PubMed: 21219646]
72. Gevers D, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe.
2014; 15:382–392. [PubMed: 24629344]
73. Chiodini RJ, et al. Microbial population differentials between mucosal and submucosal intestinal
tissues in advanced Crohn’s disease of the ileum. PLoS ONE. 2015; 10:e0134382. [PubMed:
26222621]
74. Lupp C, et al. Host-mediated inflammation disrupts the intestinal microbiota and promotes the
overgrowth of Enterobacteriaceae. Cell Host Microbe. 2:119–129. corrigendum 2, 204 (2007).
This paper reported that the induction of inflammation in the host disrupts the microbiota and
promotes a bloom of Enterobacteriaceae.
75. Garrett WS, et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous
and maternally transmitted colitis. Cell Host Microbe. 2010; 8:292–300. [PubMed: 20833380]
Author Manuscript

76. Raffatellu M, et al. Simian immunodeficiency virus-induced mucosal interleukin-17 deficiency


promotes Salmonella dissemination from the gut. Nature Med. 2008; 14:421–428. [PubMed:
18376406]
77. Godinez I, et al. T cells help to amplify inflammatory responses induced by Salmonella enterica
serotype Typhimurium in the intestinal mucosa. Infect Immun. 2008; 76:2008–2017. [PubMed:
18347048]
78. Pickard JM, et al. Rapid fucosylation of intestinal epithelium sustains host–commensal symbiosis
in sickness. Nature. 2014; 514:638–641. This paper showed that cytokines that are induced by
enteric infection promote fucosylation in the host. [PubMed: 25274297]
79. Pham TA, et al. Epithelial IL-22RA1-mediated fucosylation promotes intestinal colonization
resistance to an opportunistic pathogen. Cell Host Microbe. 2014; 16:504–516. [PubMed:
25263220]
80. Sonnenburg JL, et al. Glycan foraging in vivo by an intestine-adapted bacterial symbiont. Science.
2005; 307:1955–1959. [PubMed: 15790854]
81. Ansong C, et al. Studying Salmonellae and Yersiniae host–pathogen interactions using
Author Manuscript

integrated ’omics and modeling. Curr Top Microbiol Immunol. 2013; 363:21–41. [PubMed:
22886542]
82. Harper RW, et al. Differential regulation of dual NADPH oxidases/peroxidases, Duox1 and Duox2,
by Th1 and Th2 cytokines in respiratory tract epithelium. FEBS Lett. 2005; 579:4911–4917.
[PubMed: 16111680]
83. Haberman Y, et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and
microbiome signature. J Clin Invest. 2014; 124:3617–3633. [PubMed: 25003194]
84. Salzman A, et al. Induction and activity of nitric oxide synthase in cultured human intestinal
epithelial monolayers. Am J Physiol. 1996; 270:G565–G573. [PubMed: 8928785]

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 17

85. Palmer RM, Rees DD, Ashton DS, Moncada S. L-arginine is the physiological precursor for the
formation of nitric oxide in endothelium-dependent relaxation. Biochem Biophys Res Commun.
Author Manuscript

1988; 153:1251–1256. [PubMed: 3390182]


86. Lundberg JO, Weitzberg E, Lundberg JM, Alving K. Intragastric nitric oxide production in
humans: measurements in expelled air. Gut. 1994; 35:1543–1546. [PubMed: 7828969]
87. Singer II, et al. Expression of inducible nitric oxide synthase and nitrotyrosine in colonic
epithelium in inflammatory bowel disease. Gastroenterology. 1996; 111:871–885. [PubMed:
8831582]
88. Enocksson A, Lundberg J, Weitzberg E, Norrby-Teglund A, Svenungsson B. Rectal nitric oxide gas
and stool cytokine levels during the course of infectious gastroenteritis. Clin Diagn Lab Immunol.
2004; 11:250–254. [PubMed: 15013971]
89. Bai P, et al. Protein tyrosine nitration and poly(ADP-ribose) polymerase activation in N-methyl-N-
nitro-N-nitrosoguanidine-treated thymocytes: implication for cytotoxicity. Toxicol Lett. 2007;
170:203–213. [PubMed: 17428624]
90. Dudhgaonkar SP, Tandan SK, Kumar D, Raviprakash V, Kataria M. Influence of simultaneous
inhibition of cyclooxygenase-2 and inducible nitric oxide synthase in experimental colitis in rats.
Author Manuscript

Inflammopharmacology. 2007; 15:188–195. [PubMed: 17943250]


91. Winter SE, et al. Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science. 2013;
339:708–711. This study revealed that nitrate respiration drives the expansion of commensal E.
coli during colitis. [PubMed: 23393266]
92. Winter SE, Baumler AJ. Why related bacterial species bloom simultaneously in the gut: principles
underlying the ‘like will to like’ concept. Cell Microbiol. 2014; 16:179–184. [PubMed: 24286560]
93. Rivera-Chávez F, Baumler AJ. The pyromaniac inside you: Salmonella metabolism in the host gut.
Annu Rev Microbiol. 2015; 69:31–48. [PubMed: 26002180]
94. Galán JE, Curtiss R III. Cloning and molecular characterization of genes whose products allow
Salmonella typhimurium to penetrate tissue culture cells. Proc Natl Acad Sci USA. 1989;
86:6383–6387. [PubMed: 2548211]
95. Hensel M, et al. Simultaneous identification of bacterial virulence genes by negative selection.
Science. 1995; 269:400–403. [PubMed: 7618105]
96. Tsolis RM, Adams LG, Ficht TA, Baumler AJ. Contribution of Salmonella typhimurium virulence
Author Manuscript

factors to diarrheal disease in calves. Infect Immun. 1999; 67:4879–4885. [PubMed: 10456944]
97. Barthel M, et al. Pretreatment of mice with streptomycin provides a Salmonella enterica serovar
Typhimurium colitis model that allows analysis of both pathogen and host. Infect Immun. 2003;
71:2839–2858. [PubMed: 12704158]
98. Coburn B, Li Y, Owen D, Vallance BA, Finlay BB. Salmonella enterica serovar Typhimurium
pathogenicity island 2 is necessary for complete virulence in a mouse model of infectious
enterocolitis. Infect Immun. 2005; 73:3219–3227. [PubMed: 15908346]
99. Stecher B, et al. Salmonella enterica serovar Typhimurium exploits inflammation to compete with
the intestinal microbiota. PLoS Biol. 2007; 5:2177–2189. [PubMed: 17760501]
100. Lawley TD, et al. Host transmission of Salmonella enterica serovar Typhimurium is controlled by
virulence factors and indigenous intestinal microbiota. Infect Immun. 2008; 76:403–416.
[PubMed: 17967858]
101. Lopez CA, et al. Phage-mediated acquisition of a type III secreted effector protein boosts growth
of Salmonella by nitrate respiration. mBio. 2012; 3:e00143–12. [PubMed: 22691391]
102. Lopez CA, Rivera-Chavez F, Byndloss MX, Baumler AJ. The periplasmic nitrate reductase
Author Manuscript

NapABC supports luminal growth of Salmonella enterica serovar Typhimurium during colitis.
Infect Immun. 2015; 83:3470–3478. [PubMed: 26099579]
103. Levitt MD, Furne J, Springfield J, Suarez F, DeMaster E. Detoxification of hydrogen sulfide and
methanethiol in the cecal mucosa. J Clin Invest. 1999; 104:1107–1114. [PubMed: 10525049]
104. Harris JC, Dupont HL, Hornick RB. Fecal leukocytes in diarrheal illness. Ann Intern Med. 1972;
76:697–703. [PubMed: 4554412]
105. Loetscher Y, et al. Salmonella transiently reside in luminal neutrophils in the inflamed gut. PLoS
ONE. 2012; 7:e34812. [PubMed: 22493718]

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 18

106. Winter SE, et al. Gut inflammation provides a respiratory electron acceptor for Salmonella.
Nature. 2010; 467:426–429. This paper showed that tetrathionate is a unique electron acceptor
Author Manuscript

that is induced through inflammation that promotes expansion of S. Typhimurium. [PubMed:


20864996]
107. Muller HJ. Partial list of biological institutes and biologists doing experimental work in Russia at
the present time. Science. 1923; 57:472–473. [PubMed: 17783219]
108. Raffatellu M, et al. Lipocalin-2 resistance confers an advantage to Salmonella enterica serotype
Typhimurium for growth and survival in the inflamed intestine. Cell Host Microbe. 2009; 5:476–
486. [PubMed: 19454351]
109. Behnsen J, et al. The cytokine IL-22 promotes pathogen colonization by suppressing related
commensal bacteria. Immunity. 2014; 40:262–273. [PubMed: 24508234]
110. Goetz DH, et al. The neutrophil lipocalin NGAL is a bacteriostatic agent that interferes with
siderophore-mediated iron acquisition. Mol Cell. 2002; 10:1033–1043. [PubMed: 12453412]
111. Flo TH, et al. Lipocalin 2 mediates an innate immune response to bacterial infection by
sequestrating iron. Nature. 2004; 432:917–921. [PubMed: 15531878]
112. Deriu E, et al. Probiotic bacteria reduce Salmonella typhimurium intestinal colonization by
Author Manuscript

competing for iron. Cell Host Microbe. 2013; 14:26–37. This paper revealed that probiotic strains
of E. coli restrict infection of the gut with S. Typhimurium by competing for sources of iron.
[PubMed: 23870311]
113. Cascales E, et al. Colicin biology. Microbiol Mol Biol Rev. 2007; 71:158–229. [PubMed:
17347522]
114. Guterman SK. Colicin B: mode of action and inhibition by enterochelin. J Bacteriol. 1973;
114:1217–1224. [PubMed: 4576402]
115. Cardelli J, Konisky J. Isolation and characterization of an Escherichia coli mutant tolerant to
colicins Ia and Ib. J Bacteriol. 1974; 119:379–385. [PubMed: 4604639]
116. Patzer SI, Baquero MR, Bravo D, Moreno F, Hantke K. The colicin G, H and X determinants
encode microcins M and H47, which might utilize the catecholate siderophore receptors FepA,
Cir, Fiu and IroN. Microbiology. 2003; 149:2557–2570. [PubMed: 12949180]
117. Hooper LV, et al. Molecular analysis of commensal host–microbial relationships in the intestine.
Science. 2001; 291:881–884. This was the first report to show that the microbiota can change the
Author Manuscript

expression of mammalian genes to modulate the immune system of the host. [PubMed:
11157169]
118. Wu M, et al. Genetic determinants of in vivo fitness and diet responsiveness in multiple human
gut Bacteroides. Science. 2015; 350:aac5992. [PubMed: 26430127]
119. Earle KA, et al. Quantitative imaging of gut microbiota spatial organization. Cell Host Microbe.
2015; 18:478–488. [PubMed: 26439864]
120. Bouslimani A, et al. Molecular cartography of the human skin surface in 3D. Proc Natl Acad Sci
USA. 2015; 112:E2120–E2129. [PubMed: 25825778]
121. Marcobal A, et al. A metabolomic view of how the human gut microbiota impacts the host
metabolome using humanized and gnotobiotic mice. ISME J. 2013; 7:1933–1943. [PubMed:
23739052]
122. Lau W, Fischbach MA, Osbourn A, Sattely ES. Key applications of plant metabolic engineering.
PLoS Biol. 2014; 12:e1001879. [PubMed: 24915445]
123. Marcobal A, et al. Metabolome progression during early gut microbial colonization of gnotobiotic
mice. Sci Rep. 2015; 5:11589. [PubMed: 26118551]
Author Manuscript

124. Rath CM, et al. Molecular analysis of model gut microbiotas by imaging mass spectrometry and
nanodesorption electrospray ionization reveals dietary metabolite transformations. Anal Chem.
2012; 84:9259–9267. This study used imaging mass spectrometry to map differences in the gut
metabolic landscape that are promoted by the microbiota. [PubMed: 23009651]
125. Dorrestein PC, Mazmanian SK, Knight R. Finding the missing links among metabolites,
microbes, and the host. Immunity. 2014; 40:824–832. [PubMed: 24950202]
126. Antunes LC, et al. Antivirulence activity of the human gut metabolome. mBio. 2014; 5:e01183–
14. [PubMed: 25073640]

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 19

127. Antunes LC, et al. Impact of Salmonella infection on host hormone metabolism revealed by
metabolomics. Infect Immun. 2011; 79:1759–1769. [PubMed: 21321075]
Author Manuscript

128. Buffie CG, et al. Precision microbiome reconstitution restores bile acid mediated resistance to
Clostridium difficile. Nature. 2015; 517:205–208. This paper showed that the introduction of a
single species of bacteria that can produce secondary bile salts confers the host with resistance to
the expansion of C. difficile after treatment with antibiotics. [PubMed: 25337874]
129. Koenigsknecht MJ, Young VB. Faecal microbiota transplantation for the treatment of recurrent
Clostridium difficile infection: current promise and future needs. Curr Opin Gastroenterol. 2013;
29:628–632. [PubMed: 24100717]
130. Sekirov I, et al. Antibiotic-induced perturbations of the intestinal microbiota alter host
susceptibility to enteric infection. Infect Immun. 2008; 76:4726–4736. [PubMed: 18678663]
131. Louis E, Libioulle C, Reenaers C, Belaiche J, Georges M. Genomics of inflammatory bowel
diseases: basis for a new molecular classification and new therapeutic strategies of these diseases
[in French]. Rev Med Liege. 2009; 64:S1, 24–28.
132. Meynell GG. Antibacterial mechanisms of the mouse gut. II. The role of Eh and volatile fatty
acids in the normal gut. Br J Exp Pathol. 1963; 44:209–219. [PubMed: 13935361]
Author Manuscript

133. Donohoe DR, Wali A, Brylawski BP, Bultman SJ. Microbial regulation of glucose metabolism
and cell-cycle progression in mammalian colonocytes. PLoS ONE. 2012; 7:e46589. [PubMed:
23029553]
134. Atarashi K, Umesaki Y, Honda K. Microbiotal influence on T cell subset development. Semin
Immunol. 2011; 23:146–153. [PubMed: 21292500]
135. Atarashi K, et al. Treg induction by a rationally selected mixture of Clostridia strains from the
human microbiota. Nature. 2013; 500:232–236. [PubMed: 23842501]
136. Furusawa Y, et al. Commensal microbe-derived butyrate induces the differentiation of colonic
regulatory T cells. Nature. 2013; 504:446–450. [PubMed: 24226770]
137. Spees AM, et al. Streptomycin-induced inflammation enhances Escherichia coli gut colonization
through nitrate respiration. mBio. 2013; 4:e00430–13.
138. Itoh K, Freter R. Control of Escherichia coli populations by a combination of indigenous
clostridia and lactobacilli in gnotobiotic mice and continuous-flow cultures. Infect Immun. 1989;
57:559–565. [PubMed: 2643576]
Author Manuscript

139. Wells JE, Hylemon PB. Identification and characterization of a bile acid 7α-dehydroxylation
operon in Clostridium sp. strain TO-931, a highly active 7α-dehydroxylating strain isolated from
human feces. Appl Environ Microbiol. 2000; 66:1107–1113. [PubMed: 10698778]
140. Wilson KH. Efficiency of various bile salt preparations for stimulation of Clostridium difficile
spore germination. J Clin Microbiol. 1983; 18:1017–1019. [PubMed: 6630458]
141. Sorg JA, Sonenshein AL. Bile salts and glycine as cogerminants for Clostridium difficile spores. J
Bacteriol. 2008; 190:2505–2512. [PubMed: 18245298]
142. Theriot CM, et al. Antibiotic-induced shifts in the mouse gut microbiome and metabolome
increase susceptibility to Clostridium difficile infection. Nature Commun. 2014; 5:3114.
[PubMed: 24445449]
143. Weingarden AR, et al. Microbiota transplantation restores normal fecal bile acid composition in
recurrent Clostridium difficile infection. Am J Physiol Gastrointest Liver Physiol. 2014;
306:G310–G319. [PubMed: 24284963]
144. Bernstein H, Bernstein C, Payne CM, Dvorakova K, Garewal H. Bile acids as carcinogens in
human gastrointestinal cancers. Mutat Res. 2005; 589:47–65. [PubMed: 15652226]
Author Manuscript

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 20
Author Manuscript
Author Manuscript
Author Manuscript

Figure 1. The impact of antibiotics on the microbiota and the expansion of enteric pathogens
a, A diverse and non-disturbed microbiota confers resistance to colonization by enteric
pathogens in the intestinal epithelium. b, Treatment with antibiotics decreases the diversity
of the microbiota and leads to expansion of the C. difficile population. Toxins that are
Author Manuscript

released from C. difficile (TcdA and TcdB) enter and damage the cells of the epithelium,
which leads to inflammation (colitis) and cell death. c, Treatment with antibiotics also leads
to an increase in the levels of free sialic acid (from the host) and succinate (from the
microbiota) in the lumen of the intestine. Elevated sialic acid promotes the expansion of the
S. Typhimurium population, which can lead to inflammation (gastroenteritis) if the
bacterium invades the cells of the intestinal epithelium. Elevated levels of sialic acid and
succinate further promote the expansion of the C. difficile population and the development
of colitis and cell death.

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 21
Author Manuscript
Author Manuscript

Figure 2. Modulation of enterohaemorrhagic E. coli virulence through nutrients provided by the


microbiota
a, The microbiota resides in the lumen and outer mucus layer of the intestine. The
saccharolytic bacterium Bacteroides thetaiotaomicron is a prominent member of the
microbiota. It can release fucose from the mucus and makes the sugar available to other
bacteria. When EHEC senses fucose through the FusKR signalling system, it represses both
its use of the sugar and the expression of genes that encode the T3SS, a protein-translocation
apparatus that enables the bacterium to secrete effector proteins into host cells. This
repression prevents EHEC from competing for fucose with commensal E. coli and from
expending energy unnecessarily on T3SS expression. b, Metabolites that are provided by B.
thetaiotaomicron, such as succinate, lead to an increase in the expression by EHEC of the
enzyme mucinase, which obliterates the mucus layers of the intestine. EHEC is then able to
Author Manuscript

reach the intestinal epithelium. B. thetaiotaomicron then begins to secrete succinate and
other metabolites that are required for gluconeogenesis into the now nutrient-poor
environment. The compounds are sensed by EHEC, which upregulates its expression of the
T3SS to enable the bacterium to attach to the epithelial cells of the host intestine and form
lesions that cause diarrhoea.
Author Manuscript

Nature. Author manuscript; available in PMC 2016 November 18.


Bäumler and Sperandio Page 22
Author Manuscript
Author Manuscript

Figure 3. The effect of intestinal inflammation on nutrient availability


S. Typhimurium uses its virulence factors (T3SS-1 and T3SS-2) to trigger intestinal
inflammation. Cytokines that are released during inflammation, such as IL-22 and IFN-γ,
Author Manuscript

trigger the release of antimicrobial molecules lipocalin-2, reactive oxygen species (ROS) and
reactive nitrogen species (RNS) from the intestinal epithelium. Lipocalin-2 can block the
growth of commensal Enterobacteriaceae that rely on the siderophore enterobactin for the
acquisition of iron (Fe3+). It does not bind to the S. Typhimurium siderophone salmochelin,
however, which confers the bacterium with resistance to its effects on growth. RNS and ROS
react to form nitrate, which drives the growth of Enterobacteriaceae through nitrate
respiration. Microbiota-derived hydrogen sulfide is converted to thiosulfate by colonic
epithelial cells. Neutrophils that migrate into the lumen of the intestine during inflammation
generate ROS that convert endogenous sulfur compounds (thiosulfate) into an electron
acceptor (tetrathionate) that further boosts the growth of S. Typhimurium through
tetrathionate respiration.
Author Manuscript

Nature. Author manuscript; available in PMC 2016 November 18.

You might also like